Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Endocrinol (Oxf) ; 90(1): 192-199, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30291728

RESUMEN

OBJECTIVE: Radioiodine (RAI) is an effective treatment for Graves' thyrotoxicosis but is associated with a failure rate of 15% and may be a risk factor for thyroid eye disease (TED) and weight gain. We sought to examine predictors of RAI failure, weight gain, TED and patient satisfaction. DESIGN: Retrospective cohort study. PATIENTS: A total of 655 episodes of RAI in Graves' thyrotoxicosis patients (2006-2015). MEASUREMENTS: Biochemical assessment, including TFTs and thyrotropin receptor antibodies (TRAb), clinical features (eg, TED, weight and thionamide use) and patient questionnaire. RESULTS: The treatment failure rate was 17%. Failure was greater with higher fT4 (P = 0.002) and higher TRAb (P = 0.004). Failure rate was 42.2% when TRAb >40 U/L. Median weight gain was 3.2 kg in those with normal fT4 prior to RAI and 5.8 kg when fT4 was elevated (P < 0.001). New TED developed in 7.6% but was not associated with post-RAI dysthyroidism. Treatment satisfaction was generally high (median response 8/10). CONCLUSIONS: Treatment failure after RAI occurs in predictable groups and this should be reflected in the information provided to patients. Weight gain is common and may not entirely be explained by a return to pre-thyrotoxic baseline. We were unable to detect any significant impact of post-RAI dysthyroidism on weight gain, TED or thyroid symptoms in this large cohort.


Asunto(s)
Radioisótopos de Yodo/efectos adversos , Tirotoxicosis/radioterapia , Adulto , Estudios de Cohortes , Femenino , Oftalmopatía de Graves/etiología , Humanos , Hipotiroidismo/etiología , Radioisótopos de Yodo/uso terapéutico , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tirotoxicosis/complicaciones , Tirotoxicosis/diagnóstico , Resultado del Tratamiento , Aumento de Peso
2.
Proc Natl Acad Sci U S A ; 113(22): 6265-70, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27185937

RESUMEN

Fetoplacental glucocorticoid overexposure is a significant mechanism underlying fetal growth restriction and the programming of adverse health outcomes in the adult. Placental glucocorticoid inactivation by 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) plays a key role. We previously discovered that Hsd11b2(-/-) mice, lacking 11ß-HSD2, show marked underdevelopment of the placental vasculature. We now explore the consequences for fetal cardiovascular development and whether this is reversible. We studied Hsd11b2(+/+), Hsd11b2(+/-), and Hsd11b2(-/-) littermates from heterozygous (Hsd11b(+/-)) matings at embryonic day (E)14.5 and E17.5, where all three genotypes were present to control for maternal effects. Using high-resolution ultrasound, we found that umbilical vein blood velocity in Hsd11b2(-/-) fetuses did not undergo the normal gestational increase seen in Hsd11b2(+/+) littermates. Similarly, the resistance index in the umbilical artery did not show the normal gestational decline. Surprisingly, given that 11ß-HSD2 absence is predicted to initiate early maturation, the E/A wave ratio was reduced at E17.5 in Hsd11b2(-/-) fetuses, suggesting impaired cardiac function. Pravastatin administration from E6.5, which increases placental vascular endothelial growth factor A and, thus, vascularization, increased placental fetal capillary volume, ameliorated the aberrant umbilical cord velocity, normalized fetal weight, and improved the cardiac function of Hsd11b2(-/-) fetuses. This improved cardiac function occurred despite persisting indications of increased glucocorticoid exposure in the Hsd11b2(-/-) fetal heart. Thus, the pravastatin-induced enhancement of fetal capillaries within the placenta and the resultant hemodynamic changes correspond with restored fetal cardiac function. Statins may represent a useful therapeutic approach to intrauterine growth retardation due to placental vascular hypofunction.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/fisiología , Retardo del Crecimiento Fetal/prevención & control , Glucocorticoides/metabolismo , Cardiopatías/prevención & control , Enfermedades Placentarias/prevención & control , Pravastatina/farmacología , Animales , Anticolesterolemiantes/farmacología , Femenino , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Cardiopatías/metabolismo , Cardiopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Placentarias/metabolismo , Enfermedades Placentarias/patología , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Brain Behav Immun ; 69: 223-234, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29162555

RESUMEN

Chronically elevated glucocorticoid levels impair cognition and are pro-inflammatory in the brain. Deficiency or inhibition of 11ß-hydroxysteroid dehydrogenase type-1 (11ß-HSD1), which converts inactive into active glucocorticoids, protects against glucocorticoid-associated chronic stress- or age-related cognitive impairment. Here, we hypothesised that 11ß-HSD1 deficiency attenuates the brain cytokine response to inflammation. Because inflammation is associated with altered energy metabolism, we also examined the effects of 11ß-HSD1 deficiency upon hippocampal energy metabolism. Inflammation was induced in 11ß-HSD1 deficient (Hsd11b1Del/Del) and C57BL/6 control mice by intraperitoneal injection of lipopolysaccharide (LPS). LPS reduced circulating neutrophil and monocyte numbers and increased plasma corticosterone levels equally in C57BL/6 and Hsd11b1Del/Del mice, suggesting a similar peripheral inflammatory response. However, the induction of pro-inflammatory cytokine mRNAs in the hippocampus was attenuated in Hsd11b1Del/Del mice. Principal component analysis of mRNA expression revealed a distinct metabolic response to LPS in hippocampus of Hsd11b1Del/Del mice. Expression of Pfkfb3 and Ldha, key contributors to the Warburg effect, showed greater induction in Hsd11b1Del/Del mice. Consistent with increased glycolytic flux, levels of 3-phosphoglyceraldehyde and dihydroxyacetone phosphate were reduced in hippocampus of LPS injected Hsd11b1Del/Del mice. Expression of Sdha and Sdhb, encoding subunits of succinate dehydrogenase/complex II that determines mitochondrial reserve respiratory capacity, was induced specifically in hippocampus of LPS injected Hsd11b1Del/Del mice, together with increased levels of its product, fumarate. These data suggest 11ß-HSD1 deficiency attenuates the hippocampal pro-inflammatory response to LPS, associated with increased capacity for aerobic glycolysis and mitochondrial ATP generation. This may provide better metabolic support and be neuroprotective during systemic inflammation or aging.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Metabolismo Energético/fisiología , Hipocampo/metabolismo , Inflamación/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Corticosterona/sangre , Hipocampo/efectos de los fármacos , Conducta de Enfermedad/efectos de los fármacos , Conducta de Enfermedad/fisiología , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Monocitos/metabolismo , Neutrófilos/metabolismo
4.
Circulation ; 133(14): 1360-70, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26951843

RESUMEN

BACKGROUND: The hypertensive syndrome of Apparent Mineralocorticoid Excess is caused by loss-of-function mutations in the gene encoding 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2), allowing inappropriate activation of the mineralocorticoid receptor by endogenous glucocorticoid. Hypertension is attributed to sodium retention in the distal nephron, but 11ßHSD2 is also expressed in the brain. However, the central contribution to Apparent Mineralocorticoid Excess and other hypertensive states is often overlooked and is unresolved. We therefore used a Cre-Lox strategy to generate 11ßHSD2 brain-specific knockout (Hsd11b2.BKO) mice, measuring blood pressure and salt appetite in adults. METHODS AND RESULTS: Basal blood pressure, electrolytes, and circulating corticosteroids were unaffected in Hsd11b2.BKO mice. When offered saline to drink, Hsd11b2.BKO mice consumed 3 times more sodium than controls and became hypertensive. Salt appetite was inhibited by spironolactone. Control mice fed the same daily sodium intake remained normotensive, showing the intrinsic salt resistance of the background strain. Dexamethasone suppressed endogenous glucocorticoid and abolished the salt-induced blood pressure differential between genotypes. Salt sensitivity in Hsd11b2.BKO mice was not caused by impaired renal sodium excretion or volume expansion; pressor responses to phenylephrine were enhanced and baroreflexes impaired in these animals. CONCLUSIONS: Reduced 11ßHSD2 activity in the brain does not intrinsically cause hypertension, but it promotes a hunger for salt and a transition from salt resistance to salt sensitivity. Our data suggest that 11ßHSD2-positive neurons integrate salt appetite and the blood pressure response to dietary sodium through a mineralocorticoid receptor-dependent pathway. Therefore, central mineralocorticoid receptor antagonism could increase compliance to low-sodium regimens and help blood pressure management in cardiovascular disease.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Ansia/fisiología , Hipertensión/genética , Síndrome de Exceso Aparente de Mineralocorticoides/fisiopatología , Proteínas del Tejido Nervioso/deficiencia , Receptores de Mineralocorticoides/fisiología , Cloruro de Sodio Dietético/toxicidad , Núcleo Solitario/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/fisiología , Animales , Barorreflejo/efectos de los fármacos , Corticosterona/sangre , Dexametasona/farmacología , Conducta de Ingestión de Líquido , Genes Sintéticos , Hipertensión/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Exceso Aparente de Mineralocorticoides/tratamiento farmacológico , Síndrome de Exceso Aparente de Mineralocorticoides/genética , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Nefronas/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Nestina/genética , Neuronas/fisiología , Potasio/orina , ARN Mensajero/biosíntesis , Reflejo Anormal , Núcleo Solitario/fisiopatología , Espironolactona/farmacología
5.
Am J Epidemiol ; 185(5): 317-328, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28158597

RESUMEN

Earlier puberty, especially in girls, is associated with physical and mental disorders. Prenatal glucocorticoid exposure influences the timing of puberty in animal models, but the human relevance of those findings is unknown. We studied whether voluntary consumption of licorice, which contains glycyrrhizin (a potent inhibitor of placental 11ß-hydroxysteroid dehydrogenase type 2, the "barrier" to maternal glucocorticoids), by pregnant women was associated with pubertal maturation (height, weight, body mass index for age, difference between current and expected adult height, Tanner staging, score on the Pubertal Development Scale), neuroendocrine function (diurnal salivary cortisol, dexamethasone suppression), cognition (neuropsychological tests), and psychiatric problems (as measured by the Child Behavior Checklist) in their offspring. The children were born in 1998 in Helsinki, Finland, and examined during 2009-2011 (mean age = 12.5 (standard deviation (SD), 0.4) years; n = 378). Girls exposed to high maternal glycyrrhizin consumption (≥500 mg/week) were taller (mean difference (MD) = 0.4 SD, 95% confidence interval (CI): 0.1, 0.8), were heavier (MD = 0.6 SD, 95% CI: 0.2, 1.9), and had higher body mass index for age (MD = 0.6 SD, 95% CI: 0.2, 0.9). They were also 0.5 standard deviations (95% CI: 0.2, 0.8) closer to adult height and reported more advanced pubertal development (P < 0.04). Girls and boys exposed to high maternal glycyrrhizin consumption scored 7 (95% CI: 3.1, 11.2) points lower on tests of intelligence quotient, had poorer memory (P < 0.04), and had 3.3-fold (95% CI: 1.4, 7.7) higher odds of attention deficit/hyperactivity disorder problems compared with children whose mothers consumed little to no glycyrrhizin (≤249 mg/week). No differences in cortisol levels were found. Licorice consumption during pregnancy may be associated with harm for the developing offspring.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/inducido químicamente , Glycyrrhiza/efectos adversos , Ácido Glicirrínico/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Inteligencia/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Maduración Sexual/efectos de los fármacos , Adolescente , Antiinflamatorios/efectos adversos , Antiinflamatorios/farmacología , Índice de Masa Corporal , Niño , Factores de Confusión Epidemiológicos , Dexametasona/administración & dosificación , Dexametasona/farmacología , Femenino , Finlandia , Estudios de Seguimiento , Ácido Glicirrínico/efectos adversos , Humanos , Masculino , Embarazo , Saliva/química , Distribución por Sexo
6.
Proc Natl Acad Sci U S A ; 111(10): 3817-22, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24569863

RESUMEN

Insulin resistance and associated metabolic sequelae are common in chronic kidney disease (CKD) and are positively and independently associated with increased cardiovascular mortality. However, the pathogenesis has yet to be fully elucidated. 11ß-Hydroxysteroid dehydrogenase type 1 (11ßHSD1) catalyzes intracellular regeneration of active glucocorticoids, promoting insulin resistance in liver and other metabolic tissues. Using two experimental rat models of CKD (subtotal nephrectomy and adenine diet) which show early insulin resistance, we found that 11ßHSD1 mRNA and protein increase in hepatic and adipose tissue, together with increased hepatic 11ßHSD1 activity. This was associated with intrahepatic but not circulating glucocorticoid excess, and increased hepatic gluconeogenesis and lipogenesis. Oral administration of the 11ßHSD inhibitor carbenoxolone to uremic rats for 2 wk improved glucose tolerance and insulin sensitivity, improved insulin signaling, and reduced hepatic expression of gluconeogenic and lipogenic genes. Furthermore, 11ßHSD1(-/-) mice and rats treated with a specific 11ßHSD1 inhibitor (UE2316) were protected from metabolic disturbances despite similar renal dysfunction following adenine experimental uremia. Therefore, we demonstrate that elevated hepatic 11ßHSD1 is an important contributor to early insulin resistance and dyslipidemia in uremia. Specific 11ßHSD1 inhibitors potentially represent a novel therapeutic approach for management of insulin resistance in patients with CKD.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Resistencia a la Insulina/fisiología , ARN Mensajero/metabolismo , Insuficiencia Renal Crónica/complicaciones , Uremia/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Análisis de Varianza , Animales , Glucemia , Carbenoxolona/administración & dosificación , Carbenoxolona/farmacología , Corticosterona/sangre , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Glucocorticoides/metabolismo , Immunoblotting , Insulina/sangre , Hígado/metabolismo , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Uremia/etiología
7.
J Neurosci ; 35(2): 666-77, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25589761

RESUMEN

Maternal social stress during late pregnancy programs hypothalamo-pituitary-adrenal (HPA) axis hyper-responsiveness to stressors, such that adult prenatally stressed (PNS) offspring display exaggerated HPA axis responses to a physical stressor (systemic interleukin-1ß; IL-1ß) in adulthood, compared with controls. IL-1ß acts via a noradrenergic relay from the nucleus tractus solitarii (NTS) to corticotropin releasing hormone neurons in the paraventricular nucleus (PVN). Neurosteroids can reduce HPA axis responses, so allopregnanolone and 3ß-androstanediol (3ß-diol; 5α-reduced metabolites of progesterone and testosterone, respectively) were given subacutely (over 24 h) to PNS rats to seek reversal of the "programmed" hyper-responsive HPA phenotype. Allopregnanolone attenuated ACTH responses to IL-1ß (500 ng/kg, i.v.) in PNS females, but not in PNS males. However, 3ß-diol normalized HPA axis responses to IL-1ß in PNS males. Impaired testosterone and progesterone metabolism or increased secretion in PNS rats was indicated by greater plasma testosterone and progesterone concentrations in male and female PNS rats, respectively. Deficits in central neurosteroid production were indicated by reduced 5α-reductase mRNA levels in both male and female PNS offspring in the NTS, and in the PVN in males. In PNS females, adenovirus-mediated gene transfer was used to upregulate expression of 5α-reductase and 3α-hydroxysteroid dehydrogenase mRNAs in the NTS, and this normalized hyperactive HPA axis responses to IL-1ß. Thus, downregulation of neurosteroid production in the brain may underlie HPA axis hyper-responsiveness in prenatally programmed offspring, and administration of 5α-reduced steroids acutely to PNS rats overrides programming of hyperactive HPA axis responses to immune challenge in a sex-dependent manner.


Asunto(s)
Androstano-3,17-diol/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Pregnanolona/farmacología , Estrés Psicológico/tratamiento farmacológico , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/genética , 3-alfa-Hidroxiesteroide Deshidrogenasa (B-Específica)/metabolismo , Androstano-3,17-diol/uso terapéutico , Animales , Femenino , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Interleucina-1beta/farmacología , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/crecimiento & desarrollo , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/crecimiento & desarrollo , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Pregnanolona/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/crecimiento & desarrollo , Núcleo Solitario/metabolismo
8.
Lancet ; 385 Suppl 1: S81, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26312903

RESUMEN

BACKGROUND: Preterm infants are at increased risk of cardiometabolic disease in later life. Extrauterine growth restriction, catch-up growth, altered adiposity, and abnormal hypothalamic-pituitary-adrenal axis activity could be predisposing factors. Altered DNA methylation (5-methylcytosine, 5mC) might be one underlying mechanism. We hypothesised that preterm infants have altered 5mC at the linked differentially methylated region 2 (DMR2) of IGF2 and the H19 imprinting control region (H19 ICR) compared with term infants over the first year of life. METHODS: We recruited 46 preterm (range 25 weeks + 2 days' gestation to 31 + 5, mean 28 + 6) and 40 term infants (38 + 3 to 42 + 2 weeks' gestation, mean 40 + 2). Anthropometric variables including body composition were measured at term age and 3 months corrected age with air displacement plethysmography and at 1-year-corrected age with skin-fold thickness. Salivary cortisol was measured at 3 months corrected age after the physical examination. Percentage methylation (%5mC) was analysed with pyrosequencing on buccal DNA. Statistical analysis used Student's t test and multivariate linear regression. FINDINGS: Preterm infants demonstrated growth deficit early in postnatal life but had greater percentage body fat at term age (ß=5·73, p<0·001), but not at 3 months (ß=-0·28, p=0·82). Compared with term infants, preterm infants had a blunted cortisol response to physical examination (mean difference 0·38 µg/dL, p=0·024). At birth, preterm infants had a significant decrease in %5mC at DMR2 compared with term infants at birth (ß=-11·48, p<0·001) and compared with preterm infants at term-corrected age (t=3·13, p=0·01). By term-corrected age, preterm infants had decreased %5mC at both DMR2 (ß=-2·84, p=0·013) and the H19 ICR (ß=-2·31, p=0·048) compared with term infants at birth, although this difference disappeared at 1 year. Social deprivation was independently associated with decreased %5mC at DMR2 at birth (ß=-1·73, p=0·006) and term-corrected age (ß=-0·86, p=0·016) but not at 1 year (ß=-0·89, p=0·07). INTERPRETATION: Our results show that decreased %5mC accompanies the early growth deficit in preterm infants. The marked reduction in %5mC at IGF2 DMR2 in preterm infants at birth compared with term-age supports existing evidence that imprinting at secondary regions is established after fertilisation, whereas imprinting is established during gametogenesis at primary regions (H19 ICR). Both regions might be susceptible to early life stressors such as preterm birth and social deprivation. FUNDING: Chief Scientist Office of the Scottish Government.

9.
Eur J Neurosci ; 41(6): 787-92, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25614240

RESUMEN

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) locally regenerates active glucocorticoids from their inert forms thereby amplifying intracellular levels within target tissues including the brain. We previously showed greater increases in intra-hippocampal corticosterone (CORT) levels upon Y-maze testing in aged wild-type than in 11ß-HSD1(-/-) mice coinciding with impaired and intact spatial memory, respectively. Here we examined whether ageing influences 11ß-HSD1 regulation of CORT in the dorsal hippocampus under basal conditions during the diurnal cycle and following stress. Intra-hippocampal CORT levels measured by in vivo microdialysis in freely behaving wild-type mice displayed a diurnal variation with peak levels in the evening that were significantly elevated with ageing. In contrast, the diurnal rise in intra-hippocampal CORT levels was greatly diminished in 11ß-HSD1(-/-) mice and there was no rise with ageing; basal intra-hippocampal CORT levels were similar to wild-type controls. Furthermore, a short (3 min) swim stress induced a longer lasting increase in intra-hippocampal CORT levels in wild-type mice than in 11ß-HSD1(-/-) mice despite no genotypic differences in elevation of plasma CORT. These data indicate that 11ß-HSD1 activity contributes substantially to diurnal and stress-induced increases in hippocampal CORT levels. This contribution is even greater with ageing. Thus, 11ß-HSD1 inhibition may be an attractive target for treating cognitive impairments associated with stress or ageing.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/fisiología , Envejecimiento , Ritmo Circadiano , Corticosterona/fisiología , Hipocampo/fisiología , Estrés Psicológico , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Animales , Corticosterona/análisis , Hipocampo/química , Masculino , Ratones , Ratones Noqueados , Microdiálisis
10.
Biochem Biophys Res Commun ; 449(2): 229-34, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24832732

RESUMEN

24S,25-Epoxycholesterol is formed in a shunt of the mevalonate pathway that produces cholesterol. It is one of the most potent known activators of the liver X receptors and can inhibit sterol regulatory element-binding protein processing. Until recently analysis of 24S,25-epoxycholesterol at high sensitivity has been precluded by its thermal lability and lack of a strong chromophore. Here we report on the analysis of 24S,25-epoxycholesterol in rodent brain where its level was determined to be of the order of 0.4-1.4µg/g wet weight in both adult mouse and rat. For comparison the level of 24S-hydroxycholesterol in brain of both rodents was of the order of 20µg/g, while that of cholesterol in mouse was 10-20mg/g. By exploiting knockout mice for the enzyme oxysterol 7α-hydroxylase (Cyp7b1) we show that this enzymes is important for the subsequent metabolism of the 24S,25-epoxide.


Asunto(s)
Encéfalo/metabolismo , Colesterol/análogos & derivados , Animales , Colesterol/metabolismo , Familia 7 del Citocromo P450 , Femenino , Masculino , Redes y Vías Metabólicas , Ácido Mevalónico/metabolismo , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray , Esteroide Hidroxilasas/deficiencia , Esteroide Hidroxilasas/genética , Esteroles/metabolismo
11.
Clin Endocrinol (Oxf) ; 81(2): 175-82, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24443823

RESUMEN

OBJECTIVES: Low socio-economic status (SES) is associated with increased disease risk in the involved and the next generation. The effects of low maternal SES on the offspring may be initiated prenatally. We hypothesized that fetoplacental glucocorticoid exposure might mediate the links. We examined associations between maternal level of education and occupational status (used as indices of SES) and placental expression of genes involved in glucocorticoid exposure and transfer between the mother and foetus. DESIGN AND PATIENTS: Placental biopsies were obtained from 67 healthy women (age 32.2 ± 5.3 years) with singleton, term pregnancies without obstetric complications who participated in a prospective Prediction and Prevention of Preeclampsia (PREDO) study. MEASURES: Level of education was self-reported, and occupational status was extracted from hospital records. Relative glucocorticoid receptor (GR; NR3C1), mineralocorticoid receptor (MR; NR3C2) and 11-beta-hydroxysteroid dehydrogenase type 1 (HSD11B1) and 2 (HSD11B2) mRNA levels were quantified by real-time PCR. RESULTS: Placental GR and HSD11B1 expression increased with decreasing maternal education (unadjusted P-values for linear trend = 0.04 and 0.02 and adjusted P-values = 0.06 and 0.09, respectively). Mothers with primary/secondary education had 52.9% (95% CI, 6.2-99.6, P = 0.03, adjusted P = 0.05) and 79.6% (95% CI, 6.5-153.6, P = 0.03, adjusted P = 0.09) higher GR and HSD11B1 mRNA levels compared with mothers with tertiary education. There were no other significant associations. CONCLUSIONS: Lower maternal level of education is associated with increased placental GR and HSD11B1 gene expression. This combination may regenerate active glucocorticoids in placenta and increase placental sensitivity to glucocorticoids, potentially leading to greater placental and foetal glucocorticoid exposure.


Asunto(s)
Escolaridad , Empleo , Placenta/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Adulto , Femenino , Humanos , Masculino , Embarazo , Estudios Prospectivos , ARN Mensajero , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética
12.
Reprod Biol Endocrinol ; 12: 73, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25082159

RESUMEN

BACKGROUND: Maternal overnutrition during pregnancy is associated with an increased risk of obesity and cardiometabolic disease in the offspring; a phenomenon attributed to 'developmental programming'. The post-weaning development of obesity may associate with exacerbation of the programmed metabolic phenotype. In mice, we have previously shown that exposure to maternal overnutrition causes increased weight gain in offspring before weaning, but exerts no persistent effects on weight or glucose tolerance in adulthood. In order to determine whether post-weaning exposure to a cafeteria diet might lead to an exacerbation of programmed effects, offspring born and raised by mothers on control (CON) or cafeteria (DIO) diets were transferred onto either CON or DIO diets at weaning. FINDINGS: Post-weaning DIO caused the development of obesity, with hyperglycaemia and hyperinsulinaemia in males; and obesity with hyperinsulinaemia in females and with increased cholesterol levels in both sexes. Exposure to maternal overnutrition during pregnancy and lactation caused only subtle additional effects on offspring phenotype. CONCLUSIONS: These results suggest that post-weaning exposure to a high-fat high-sugar diet has a more profound effect on offspring weight gain and glucose tolerance than exposure to maternal overnutrition. These data emphasise the importance of optimising early life nutrition in offspring of both obese and lean mothers.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Sacarosa en la Dieta/efectos adversos , Resistencia a la Insulina , Síndrome Metabólico/etiología , Obesidad/etiología , Hipernutrición/fisiopatología , Animales , Desarrollo Embrionario , Femenino , Hipercolesterolemia/etiología , Hiperglucemia/etiología , Hiperinsulinismo/etiología , Lactancia , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Síndrome Metabólico/sangre , Síndrome Metabólico/fisiopatología , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/fisiopatología , Hipernutrición/etiología , Embarazo , Caracteres Sexuales , Destete , Aumento de Peso
13.
FASEB J ; 27(4): 1519-31, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23303209

RESUMEN

11ß-Hydroxysteroid dehydrogenase type-1 (11ß-HSD1) converts inert cortisone into active cortisol, amplifying intracellular glucocorticoid action. 11ß-HSD1 deficiency improves cardiovascular risk factors in obesity but exacerbates acute inflammation. To determine the effects of 11ß-HSD1 deficiency on atherosclerosis and its inflammation, atherosclerosis-prone apolipoprotein E-knockout (ApoE-KO) mice were treated with a selective 11ß-HSD1 inhibitor or crossed with 11ß-HSD1-KO mice to generate double knockouts (DKOs) and challenged with an atherogenic Western diet. 11ß-HSD1 inhibition or deficiency attenuated atherosclerosis (74-76%) without deleterious effects on plaque structure. This occurred without affecting plasma lipids or glucose, suggesting independence from classical metabolic risk factors. KO plaques were not more inflamed and indeed had 36% less T-cell infiltration, associated with 38% reduced circulating monocyte chemoattractant protein-1 (MCP-1) and 36% lower lesional vascular cell adhesion molecule-1 (VCAM-1). Bone marrow (BM) cells are key to the atheroprotection, since transplantation of DKO BM to irradiated ApoE-KO mice reduced atherosclerosis by 51%. 11ß-HSD1-null macrophages show 76% enhanced cholesterol ester export. Thus, 11ß-HSD1 deficiency reduces atherosclerosis without exaggerated lesional inflammation independent of metabolic risk factors. Selective 11ß-HSD1 inhibitors promise novel antiatherosclerosis effects over and above their benefits for metabolic risk factors via effects on BM cells, plausibly macrophages.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/deficiencia , Aterosclerosis/metabolismo , Médula Ósea/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Aterosclerosis/genética , Médula Ósea/efectos de los fármacos , Glucocorticoides/metabolismo , Ratones , Ratones Noqueados , Factores de Riesgo , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Cell Metab ; 7(4): 286-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18396133

RESUMEN

Although elevated cortisol levels may be found in diabetes, cortisol's role in diabetes-associated cognitive impairment is unclear. A recent study (Stranahan et al., 2008) shows that reducing glucocorticoid levels in rodent diabetes models prevents cognitive deficits and enhances neurogenesis and synaptic plasticity. The clinical relevance of these intriguing findings remains undetermined.


Asunto(s)
Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/fisiopatología , Complicaciones de la Diabetes , Diabetes Mellitus/fisiopatología , Glucocorticoides/metabolismo , Animales , Corticosterona/metabolismo , Humanos , Ratones , Ratas
15.
J Biol Chem ; 287(6): 4188-97, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22158867

RESUMEN

In obesity, rapidly expanding adipose tissue becomes hypoxic, precipitating inflammation, fibrosis, and insulin resistance. Compensatory angiogenesis may prevent these events. Mice lacking the intracellular glucocorticoid-amplifying enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1(-/-)) have "healthier" adipose tissue distribution and resist metabolic disease with diet-induced obesity. Here we show that adipose tissues of 11ßHSD1(-/-) mice exhibit attenuated hypoxia, induction of hypoxia-inducible factor (HIF-1α) activation of the TGF-ß/Smad3/α-smooth muscle actin (α-SMA) signaling pathway, and fibrogenesis despite similar fat accretion with diet-induced obesity. Moreover, augmented 11ßHSD1(-/-) adipose tissue angiogenesis is associated with enhanced peroxisome proliferator-activated receptor γ (PPARγ)-inducible expression of the potent angiogenic factors VEGF-A, apelin, and angiopoietin-like protein 4. Improved adipose angiogenesis and reduced fibrosis provide a novel mechanism whereby suppression of intracellular glucocorticoid regeneration promotes safer fat expansion with weight gain.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Tejido Adiposo/enzimología , Hipoxia/enzimología , Neovascularización Fisiológica , Obesidad/enzimología , Transducción de Señal , Actinas/genética , Actinas/metabolismo , Adipoquinas , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/patología , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Animales , Apelina , Fibrosis/enzimología , Fibrosis/genética , Fibrosis/fisiopatología , Hipoxia/patología , Hipoxia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Resistencia a la Insulina/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/patología , Obesidad/fisiopatología , PPAR gamma/genética , PPAR gamma/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Aumento de Peso/genética
16.
Curr Atheroscler Rep ; 15(5): 320, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23512604

RESUMEN

Atherosclerosis is a chronic inflammatory disease in which initial vascular damage leads to extensive macrophage and lymphocyte infiltration. Although acutely glucocorticoids suppress inflammation, chronic glucocorticoid excess worsens atherosclerosis, possibly by exacerbating systemic cardiovascular risk factors. However, glucocorticoid action within the lesion may reduce neointimal proliferation and inflammation. Glucocorticoid levels within cells do not necessarily reflect circulating levels due to pre-receptor metabolism by 11ß-hydroxysteroid dehydrogenases (11ß-HSDs). 11ß-HSD2 converts active glucocorticoids into inert 11-keto forms. 11ß-HSD1 catalyses the reverse reaction, regenerating active glucocorticoids. 11ß-HSD2-deficiency/inhibition causes hypertension, whereas deficiency/inhibition of 11ß-HSD1 generates a cardioprotective lipid profile and improves glycemic control. Importantly, 11ß-HSD1-deficiency/inhibition is atheroprotective, whereas 11ß-HSD2-deficiency accelerates atherosclerosis. These effects are largely independent of systemic risk factors, reflecting modulation of glucocorticoid action and inflammation within the vasculature. Here, we consider whether evidence linking the 11ß-HSDs to vascular inflammation suggests these isozymes are potential therapeutic targets in vascular injury and atherosclerosis.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/fisiología , Aterosclerosis/inmunología , Glucocorticoides/inmunología , Vasculitis/inmunología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/inmunología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/inmunología , Aterosclerosis/complicaciones , Aterosclerosis/enzimología , Glucocorticoides/metabolismo , Humanos , Neointima/inmunología , Vasculitis/complicaciones , Vasculitis/enzimología
17.
FASEB J ; 26(5): 1866-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22321728

RESUMEN

Fetal growth restriction associates with increased risk of adult cardiometabolic and neuropsychiatric disorders. Both maternal malnutrition [notably a low-protein (LP) diet] and stress/glucocorticoid exposure reduce fetal growth and cause persisting abnormalities (programming) in adult offspring. Deficiency of placental 11ß-hydroxysteroid dehydrogenase-2 (11ß-HSD2), which inactivates glucocorticoids, is reduced by an LP diet and has been proposed as a unifying mechanism. Here, we explored the importance of glucocorticoids and placental 11ß-HSD2 in dietary programming. Pregnant mice were fed a control or isocaloric LP diet throughout gestation. The LP diet first elevated fetal glucocorticoid levels, then reduced placental growth, and finally decreased fetal weight near term by 17%. Whereas the LP diet reduced placental 11ß-HSD2 activity near term by ∼25%, consistent with previous reports, activity was increased between 20 and 40% at earlier ages, implying that glucocorticoid overexposure in LP fetuses occurs via 11ß-HSD2-independent mechanisms. Consistent with this, heterozygous 11ß-HSD2(+/-) crosses showed that although both LP and 11ß-HSD2 deficiency reduced fetal growth, LP indeed acted independently of 11ß-HSD2. Instead, the LP diet induced the fetal hypothalamic-pituitary-adrenal axis per se. Thus, maternal malnutrition and placental 11ß-HSD2 deficiency act via distinct processes to retard fetal growth, both involving fetoplacental overexposure to glucocorticoids but from distinct sources.


Asunto(s)
Desarrollo Fetal , Glucocorticoides/fisiología , Estado Nutricional , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Animales , Secuencia de Bases , Corticosterona/sangre , Cartilla de ADN , Femenino , Hibridación in Situ , Exposición Materna , Ratones , Ratones Endogámicos C57BL , Placenta/enzimología , Embarazo
18.
Cerebellum ; 12(1): 68-73, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22699965

RESUMEN

The cerebellum participates in multiple cognitive functions, including those that are sensitive to decline with aging, and is also vulnerable to atrophy with aging. However, few studies have examined structure-function relationships in older adults. We measured the cross-sectional area of four areas of the cerebellar vermis in 45 community-dwelling men aged 71-76, and correlated this with individual cognitive test scores and two cognitive factors derived from principal components analysis. Two out of the four areas showed positive correlations; vermis area 4 (lobules VIII-X) correlated at r = 0.47 (p = 0.001) with a general cognitive factor accounting for almost half of the cognitive test variance. These findings support the hypothesis that variations in cerebellar structure are associated with cognitive ability in older adults.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Cerebelo/patología , Cerebelo/fisiología , Cognición/fisiología , Anciano , Atrofia/patología , Atrofia/fisiopatología , Humanos , Masculino , Memoria/fisiología , Características de la Residencia , Conducta Verbal/fisiología
19.
Arterioscler Thromb Vasc Biol ; 32(3): 677-85, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22207732

RESUMEN

OBJECTIVE: 11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) catalyzes the intracellular reduction of inactive cortisone to active cortisol, the natural ligand activating the glucocorticoid receptor (GR). Peroxisome proliferator- activated receptor-γ (PPARγ) is a nuclear receptor controlling inflammation, lipid metabolism, and the macrophage polarization state. In this study, we investigated the impact of macrophage polarization on the expression and activity of 11ß-HSD1 and the role of PPARγ therein. METHODS AND RESULTS: 11ß-HSD1 gene expression is higher in proinflammatory M1 and anti-inflammatory M2 macrophages than in resting macrophages, whereas its activity is highest in M2 macrophages. Interestingly, PPARγ activation induces 11ß-HSD1 enzyme activity in M2 macrophages but not in resting macrophages or M1 macrophages. Consequently, human M2 macrophages displayed enhanced responsiveness to the 11ß-HSD1 substrate cortisone, an effect amplified by PPARγ induction of 11ß-HSD1 activity, as illustrated by an increased expression of GR target genes. CONCLUSION: Our data identify a positive cross-talk between PPARγ and GR in human M2 macrophages via the induction of 11ß-HSD1 expression and activity.


Asunto(s)
Inflamación/enzimología , Macrófagos/efectos de los fármacos , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/biosíntesis , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Células Cultivadas , Cortisona/metabolismo , Inducción Enzimática , Genes Reporteros , Humanos , Hidrocortisona/metabolismo , Inflamación/genética , Inflamación/inmunología , Interleucina-4/metabolismo , Macrófagos/enzimología , Macrófagos/inmunología , PPAR gamma/genética , PPAR gamma/metabolismo , Interferencia de ARN , Receptores de Glucocorticoides/metabolismo , Rosiglitazona , Factores de Tiempo , Transfección
20.
J Neurosci ; 31(11): 4188-93, 2011 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-21411659

RESUMEN

Local brain amplification of glucocorticoids (GCs) by 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) plays a pivotal role in age-related memory deficits. 11ß-HSD1 deficient mice are protected from spatial memory impairments with aging, but the underlying mechanisms are unknown. To determine which brain receptors [high-affinity mineralocorticoid receptors (MRs) or low-affinity glucocorticoid receptors (GRs)] are involved, spatial memory was measured in aged 11ß-HSD1(-/-) mice before and during intracerebroventricular infusion (10 d) of spironolactone (MR antagonist) or RU486 (GR antagonist). Aged C57BL/6J control mice showed impaired spatial memory in the Y-maze; this improved with GR blockade, while MR blockade had no effect. In contrast, aged 11ß-HSD1(-/-) mice showed intact spatial memory that became impaired with MR blockade, but not GR blockade. Hippocampal MR and GR mRNA expression and plasma corticosterone levels were not significantly altered with spironolactone or RU486 in either genotype. These data support the notion that 11ß-HSD1 deficiency in aging mice leads to lower intracellular GC concentrations in brain, particularly in the hippocampus, which activate predominantly MRs to enhance memory, while in aging C57BL/6J controls, the increased intracellular GCs saturate MRs and activate predominantly GRs, thus impairing memory, an effect reversed by GR blockade.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Envejecimiento/fisiología , Hipocampo/metabolismo , Memoria/fisiología , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Análisis de Varianza , Animales , Cognición/efectos de los fármacos , Cognición/fisiología , Corticosterona/sangre , Hipocampo/efectos de los fármacos , Infusiones Intraventriculares , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria/efectos de los fármacos , Ratones , Ratones Noqueados , Antagonistas de Receptores de Mineralocorticoides/farmacología , Radioinmunoensayo , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/genética , Espironolactona/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA